Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Radiat Res ; 196(3): 284-296, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34153091

RESUMEN

Thrombocytopenia is a major complication in hematopoietic-acute radiation syndrome (H-ARS) that increases the risk of mortality from uncontrolled hemorrhage. There is a great demand for new therapies to improve survival and mitigate bleeding in H-ARS. Thrombopoiesis requires interactions between megakaryocytes (MKs) and endothelial cells. 16, 16-dimethyl prostaglandin E2 (dmPGE2), a longer-acting analogue of PGE2, promotes hematopoietic recovery after total-body irradiation (TBI), and various angiotensin-converting enzyme (ACE) inhibitors mitigate endothelial injury after radiation exposure. Here, we tested a combination therapy of dmPGE2 and lisinopril to mitigate thrombocytopenia in murine models of H-ARS following TBI. After 7.75 Gy TBI, dmPGE2 and lisinopril each increased survival relative to vehicle controls. Importantly, combined dmPGE2 and lisinopril therapy enhanced survival greater than either individual agent. Studies performed after 4 Gy TBI revealed reduced numbers of marrow MKs and circulating platelets. In addition, sublethal TBI induced abnormalities both in MK maturation and in in vitro and in vivo platelet function. dmPGE2, alone and in combination with lisinopril, improved recovery of marrow MKs and peripheral platelets. Finally, sublethal TBI transiently reduced the number of marrow Lin-CD45-CD31+Sca-1- sinusoidal endothelial cells, while combined dmPGE2 and lisinopril treatment, but not single-agent treatment, accelerated their recovery. Taken together, these data support the concept that combined dmPGE2 and lisinopril therapy improves thrombocytopenia and survival by promoting recovery of the MK lineage, as well as the MK niche, in the setting of H-ARS.


Asunto(s)
16,16-Dimetilprostaglandina E2/uso terapéutico , Síndrome de Radiación Aguda/tratamiento farmacológico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Plaquetas/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Trastornos Hemorrágicos/tratamiento farmacológico , Lisinopril/uso terapéutico , Megacariocitos/efectos de los fármacos , Trombocitopenia/tratamiento farmacológico , Trombopoyesis/efectos de los fármacos , Síndrome de Radiación Aguda/complicaciones , Animales , Plaquetas/efectos de la radiación , Médula Ósea/efectos de los fármacos , Médula Ósea/efectos de la radiación , Proteína C-Reactiva/análisis , Radioisótopos de Cesio , Evaluación Preclínica de Medicamentos , Células Endoteliales/efectos de la radiación , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/efectos de la radiación , Femenino , Rayos gamma/efectos adversos , Trastornos Hemorrágicos/etiología , Megacariocitos/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Selectina-P/análisis , Agregación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de la radiación , Factor Plaquetario 4/análisis , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/etiología , Trombocitopenia/etiología , Trombopoyesis/efectos de la radiación , Irradiación Corporal Total , Factor de von Willebrand/análisis
3.
Int J Radiat Biol ; 96(12): 1597-1607, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32990492

RESUMEN

PURPOSE: Depleted uranium (DU) has several civilian and military applications. The effects of this emerging environmental pollutant on human health raise some concerns. Previous experimental studies have shown that uranium (U) exposure can disturb the central nervous system. A small quantity of U reaches the brain via the blood, but the effects on the blood-brain barrier (BBB) remain unclear. MATERIALS AND METHODS: In the present work, two cell culture models were exposed to DU for different times to study its cytotoxicity, paracellular permeability and extracellular concentration of U. The well-known immortalized human cerebral microvascular endothelial cells, hCMEC/D3, were cultured on the filter in the first model. In the second model, human primary cells of pericytes were cultured under the filter to understand the influence of cell environment after U exposure. RESULTS: The results show that U is not cytotoxic to hCMEC/D3 cells or pericytes until 500 µM (1.6 Bq.L-1). In addition, acute or chronic low-dose exposure of U did not disturb permeability and was conserved in both cell culture models. However, U is able to reach the brain compartment. During the first hours of exposure, the passage of U to the abluminal compartment was significantly reduced in the presence of pericytes. Electronic microscopy studies evidenced the formation of needlelike structures, like urchin-shaped precipitates, from 1 h of exposure. Analytical microscopy confirmed the U composition of these precipitates. Interestingly, precipitated U was detected only in endothelial cells and not in pericytes. U was localized in multilamellar or multivesicular bodies along the endo-lysosomal pathway, suggesting the involvement of these traffic vesicles in U sequestration and/or elimination. CONCLUSIONS: We show for the first time the in vitro passage of U across a human cerebral microvascular endothelial cells, and the intracellular localization of U precipitates without any cytotoxicity or modification of paracellular permeability. The difference between the results obtained with monolayers and co-culture models with pericytes illustrates the need to use complex in vitro models in order to mimic the neurovascular unit. Further in vivo studies should be performed to better understand the passage of U across the blood-brain barrier potentially involved in behavioral consequences.


Asunto(s)
Encéfalo/irrigación sanguínea , Células Endoteliales/metabolismo , Microvasos/citología , Uranio/metabolismo , Barrera Hematoencefálica/metabolismo , Línea Celular , Técnicas de Cocultivo , Células Endoteliales/efectos de la radiación , Espacio Extracelular/metabolismo , Espacio Extracelular/efectos de la radiación , Humanos , Permeabilidad , Factores de Tiempo
4.
Lasers Med Sci ; 34(3): 495-504, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30145725

RESUMEN

Photobiomodulation of cells using near-infrared (NIR) monochromatic light can affect cell functions such as proliferation, viability, and metabolism in a range of cell types. Evidence for the effects of near-infrared light on endothelial cells has been reported, but the studies were mainly performed using VIS light emitted by low-energy lasers, because NIR wavelengths seemed negatively stimulate these cells. Cell viability, free radical-induced oxidative stress, NF-κB activation, nitric oxide release, mitochondrial respiration, and wound healing repair were assessed in human endothelial cells (HECV) irradiated with 808-nm diode laser light (laser setup = 1 W/cm2, 60 s, 60 J/cm2, CW vs measured energy parameter = 0.95 W/cm2, 60 s, 57 J/cm2, mode CW) emitted by an handpiece with flat-top profile. No difference in viability was detected between controls and HECV cells irradiated with 808-nm diode laser light for 60 s. Irradiated cells demonstrated higher proliferation rate and increased migration ability associated to moderate increase in ROS production without a significant increase in oxidative stress and oxidative stress-activated processes. Near-infrared light stimulated mitochondrial oxygen consumption and ATP synthesis in HECV cells. Short near-infrared irradiation did not affect viability of HECV cells, rather led to a stimulation of wound healing rate, likely sustained by ROS-mediated stimulation of mitochondrial activity. Our results demonstrating that near-infrared led to a shift from anaerobic to aerobic metabolism provide new insight into the possible molecular mechanisms by which photobiomodulation with 808-nm diode laser light protects against inflammation-induced endothelial dysfunction, seemingly promising to enhance their therapeutic properties.


Asunto(s)
Células Endoteliales/efectos de la radiación , Láseres de Semiconductores , Terapia por Luz de Baja Intensidad , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación , Fosforilación Oxidativa/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo , Cicatrización de Heridas/efectos de la radiación , Aerobiosis , Línea Celular , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Células Endoteliales/metabolismo , Humanos , Óxido Nítrico/metabolismo
5.
Exp Dermatol ; 27(9): 973-980, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29742305

RESUMEN

Red grape (Vitis vinifera L.) flavonoids including flavan-3-ols (eg, catechin and epicatechin), flavonols (eg, quercetin) and anthocyanins (eg, malvidin) exert anti-inflammatory and antioxidant activities. In the skin they also have a photoprotective action, and their effects have been extensively investigated in keratinocytes, melanocytes and fibroblasts. Despite their known effects also on blood vasculature, little is known on their activities on human dermal blood endothelial cells (HDBECs), which are critically involved in skin homeostasis as well as in the pathogenesis of neoplastic and inflammatory skin diseases. We sought to study the biological effects of selected red grape flavonoids in preventing the consequences of ultraviolet (UV)-A irradiation in vitro. Our results show that red grape flavonoids prevent UV-A-induced sICAM-1 release in HDBECs, suggesting that this cell type could represent an additional target of the anti-inflammatory activity of flavonoids. In addition, flavonoids effectively inhibited UV-A-induced synthesis of collagen type III at both RNA and protein level, indicating that dermal blood microvasculature could be actively involved in ECM remodelling as a consequence of skin photo-ageing, and that this can be prevented by red grape flavonoids.


Asunto(s)
Colágeno Tipo III/biosíntesis , Células Endoteliales/efectos de los fármacos , Células Endoteliales/efectos de la radiación , Flavonoides/farmacología , Extractos Vegetales/farmacología , Vitis , Catequina/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Colágeno Tipo III/genética , Frutas , Expresión Génica/efectos de los fármacos , Expresión Génica/efectos de la radiación , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Dímeros de Pirimidina/metabolismo , Quercetina/análogos & derivados , Quercetina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Piel/citología , Rayos Ultravioleta , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/efectos de la radiación
6.
Lasers Med Sci ; 33(2): 279-286, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29098460

RESUMEN

Low-level laser therapy (LLLT), widely used in physiotherapy, has been known to enhance wound healing and stimulate cell proliferation, including fibroblast and endothelial cells. Applying LLLT can increase cell proliferation in many kinds of cells including fibroblasts and endothelial cells. However, the protective mechanisms of LLLT on endothelial apoptosis remain unclear. We hypothesized LLLT can protect endothelial cells from inflammation-induced apoptosis. Human endothelial cell line, EA.hy926 cells, and TNF-α/cycloheximide (TNF/CHX) were used to explore the protective effects of LLLT (660 nm) on inflammation-induced endothelial apoptosis. Cell viability, apoptosis, caspase-3/7/8/9 activity, MAPKs signaling, NF-κB activity, and inducible/endothelial nitric oxide synthase (iNOS/eNOS) expression were measured. Our results showed that LLLT increased EA.hy926 cell proliferation, attenuated the TNF/CHX-induced apoptosis, and reduced the TNF/CHX-mediated caspase-3/7/8/9 activation. In addition, LLLT increased ERK MAPK phosphorylation and suppressed the TNF/CHX-increased p38 MAPK, JNK, IKK phosphorylation, NF-κB translocation, and iNOS expression. The caspases-3 cleavage and cell death were not increased in cells treating with ERK inhibitor U0126, which implicated that ERK is not to be responsible for the protective effects of LLLT. After treating with p38 mitogen-activated protein kinase (MAPK) activator, the protection of LLLT in cell apoptosis was no longer existed, showing that LLLT protected the endothelial cells by suppressing p38 MAPK signaling. Our results provide a new insight into the possible molecular mechanisms in which LLLT protects against inflammatory-induced endothelial dysfunction.


Asunto(s)
Apoptosis/efectos de la radiación , Cicloheximida/efectos adversos , Células Endoteliales/patología , Células Endoteliales/efectos de la radiación , Terapia por Luz de Baja Intensidad , Factor de Necrosis Tumoral alfa/efectos adversos , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Endoteliales/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/efectos de la radiación , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de la radiación , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Sci Rep ; 7(1): 10700, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28878330

RESUMEN

Low level light therapy receives increasing interest in the fields of tissue regeneration and wound healing. Several in vivo studies demonstrated the positive effects of LLLT on angiogenesis. This study aimed to investigate the underlying properties in vitro by comparing the effects of light therapy by light emitting diodes of different wavelengths on endothelial cells in vitro. Human umbilical vein endothelial cells were treated with either 475 nm, 516 nm or 635 nm light. Control cells were not illuminated. 2D proliferation was quantified by manual counting. HUVEC migration was analyzed by performing a 2D wound scratch assay and a 3D bead assay. The influence of LLLT on early vasculogenic events was determined in a 3D fibrin co-culture model with adipose-derived stem cells. Stimulation with both red and green pulsed LED light significantly increased HUVEC proliferation and 3D migration. Moreover, HUVEC showed increased 2D migration potential with green light stimulation. The treatment with blue light was ineffective. Several parameters showed that green light was even more potent to stimulate proliferation and migration of endothelial cells than clinically well-established red light therapy. Further studies have to focus on intracellular mechanisms induced by different wavelengths in order to optimize this promising therapy in tissue regeneration.


Asunto(s)
Células Endoteliales/efectos de la radiación , Luz , Fototerapia , Biomarcadores , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Células Cultivadas , Células Endoteliales/metabolismo , Expresión Génica , Genes Reporteros , Células Endoteliales de la Vena Umbilical Humana , Humanos , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo
8.
Lasers Med Sci ; 32(8): 1737-1746, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28653257

RESUMEN

Skin flap grafting is a form of transplantation widely used in plastic surgery. However, ischemia/reperfusion injury is the main factor which reduces the survival rate of flaps following grafting. We investigated whether photobiomodulation (PBM) precondition prior to human adipose-derived stromal cell (hASC) spheroid (PBM-spheroid) transplantation improved skin tissue functional recovery by the stimulation of angiogenesis and tissue regeneration in skin flap of mice. The LED had an emission wavelength peaked at 660 ± 20 nm (6 J/cm2, 10 mW/cm2). The expression of angiogenic growth factors in PBM-spheroid hASCs was much greater than that of not-PBM-treated spheroid or monolayer-cultured hASCs. From immunochemical staining analysis, the hASCs of PBM-spheroid were CD31+, KDR+, and CD34+, whereas monolayer-cultured hASCs were negative for these markers. To evaluate the therapeutic effect of hASC PBM-spheroid in vivo, PBS, monolayer-cultured hASCs, and not-PBM-spheroid were transplanted into a skin flap model. The animals were observed for 14 days. The PBM-spheroid hASCs transplanted into the skin flap ischemia differentiated into endothelial cells and remained differentiated. Transplantation of PBM-spheroid hASCs into the skin flap ischemia significantly elevated the density of vascular formations through angiogenic factors released by the skin flap ischemia and enhanced tissue regeneration at the lesion site. Consistent with these results, the transplantation of PBM-spheroid hASCs significantly improved functional recovery compared with PBS, monolayer-cultured hASCs, and not-PBM-spheroid treatment. These findings suggest that transplantation of PBM-spheroid hASCs may be an effective stem cell therapy for the treatment of skin flap ischemia.


Asunto(s)
Tejido Adiposo/citología , Isquemia/terapia , Terapia por Luz de Baja Intensidad , Regeneración/efectos de la radiación , Piel/irrigación sanguínea , Esferoides Celulares/citología , Células Madre/citología , Colgajos Quirúrgicos/irrigación sanguínea , Animales , Diferenciación Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/efectos de la radiación , Células Epiteliales/citología , Células Epiteliales/efectos de la radiación , Humanos , Isquemia/patología , Ratones , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de la radiación , Neovascularización Fisiológica/efectos de la radiación , Piel/patología , Esferoides Celulares/efectos de la radiación , Trasplante de Células Madre , Células Madre/efectos de la radiación , Cicatrización de Heridas
9.
PLoS One ; 12(4): e0176243, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28448534

RESUMEN

A wide search for ischemic preconditioning (IPC) mechanisms of cardioprotection identified the light elicited circadian rhythm protein Period 2 (Per2) to be cardioprotective. Studies on cardiac metabolism found a key role for light elicited Per2 in mediating metabolic dependence on carbohydrate metabolism. To profile Per2 mediated pathways following IPC of the mouse heart, we performed a genome array and identified 352 abundantly expressed and well-characterized Per2 dependent micro RNAs. One prominent result of our in silico analysis for cardiac Per2 dependent micro RNAs revealed a selective role for miR-21 in the regulation of hypoxia and metabolic pathways. Based on this Per2 dependency, we subsequently found a diurnal expression pattern for miR-21 with higher miR-21 expression levels at Zeitgeber time (ZT) 15 compared to ZT3. Gain or loss of function studies for miR-21 using miRNA mimics or miRNA inhibitors and a Seahorse Bioanalyzer uncovered a critical role of miR-21 for cellular glycolysis, glycolytic capacity, and glycolytic reserve. Exposing mice to intense light, a strategy to induce Per2, led to a robust induction of cardiac miR-21 tissue levels and decreased infarct sizes, which was abolished in miR-21-/- mice. Similarly, first translational studies in humans using intense blue light exposure for 5 days in healthy volunteers resulted in increased plasma miR-21 levels which was associated with increased phosphofructokinase activity, the rate-limiting enzyme in glycolysis. Together, we identified miR-21 as cardioprotective downstream target of Per2 and suggest intense light therapy as a potential strategy to enhance miR-21 activity and subsequent carbohydrate metabolism in humans.


Asunto(s)
Glucólisis/efectos de la radiación , Corazón/efectos de la radiación , Luz , MicroARNs/genética , Miocardio/metabolismo , Proteínas Circadianas Period/metabolismo , Regulación hacia Arriba/efectos de la radiación , Adulto , Animales , Células Endoteliales/metabolismo , Células Endoteliales/efectos de la radiación , Femenino , Humanos , Precondicionamiento Isquémico , Pulmón/metabolismo , Pulmón/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/patología , Proteínas Circadianas Period/deficiencia , Fosfofructoquinasas/metabolismo , Adulto Joven
10.
Lasers Med Sci ; 31(5): 1017-25, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27147074

RESUMEN

Bleeding is a common feature in envenoming caused by Bothrops snake venom due to extensive damage to capillaries and venules, producing alterations in capillary endothelial cell morphology. It has been demonstrated, in vivo, that photobiomodulation (PBM) decreases hemorrhage after venom inoculation; however, the mechanism is unknown. Thus, the objective was to investigate the effects of PBM on a murine endothelial cell line (tEnd) exposed to Bothrops jararaca venom (BjV). Cells were exposed to BjV and irradiated once with either 660- or 780-nm wavelength laser light at energy densities of 4 and 5 J/cm(2), respectively, and irradiation time of 10 s. Cell integrity was analyzed by crystal violet and cell viability/mitochondrial metabolism by MTT assay. The release of lactic dehydrogenase (LDH) was quantified as a measure of cell damage. In addition, cytokine IL1-ß levels were measured in the supernatant. PBM at 660 and 780 nm wavelength was able to increase cellular viability and decrease the release of LDH and the loss of cellular integrity. In addition, the concentration of pro-inflammatory cytokine IL1-ß was reduced after PBM by both wavelengths. The data reported herein indicates that irradiation with red or near-infrared laser resulted in protection on endothelial cells after exposure to Bothrops venom and could be, at least in part, a reasonable explanation by the beneficial effects of PBM inhibiting the local effects induced by Bothrops venoms, in vivo.


Asunto(s)
Venenos de Crotálidos/envenenamiento , Células Endoteliales/efectos de la radiación , Hemorragia/inducido químicamente , Hemorragia/radioterapia , Terapia por Luz de Baja Intensidad/métodos , Animales , Bothrops , Supervivencia Celular/efectos de la radiación , Interleucina-1beta/efectos de la radiación , Ratones
11.
Lasers Med Sci ; 31(5): 825-31, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26861982

RESUMEN

Diabetes mellitus is considered to be a very serious lifestyle disease leading to cardiovascular complications and impaired wound healing observed in the diabetic foot syndrome. Chronic hyperglycemia is the source of the endothelial activation. The inflammatory process in diabetes is associated with the secretion of inflammatory cytokines by endothelial cells, e.g., tumor necrosis factor-alpha (TNF-α) and interleukin 6 (IL-6). The method of phototherapy using laser beam of low power (LLLT-low-level laser therapy) effectively supports the conventional treatment of diabetic vascular complications such as diabetic foot syndrome. The aim of our study was to evaluate the effect of low-power laser irradiation at two wavelengths (635 and 830 nm) on the secretion of inflammatory factors (TNF-α and IL-6) by the endothelial cell culture-HUVEC line (human umbilical vein endothelial cell)-under conditions of hyperglycemia. It is considered that adverse effects of hyperglycemia on vascular endothelial cells may be corrected by the action of LLLT, especially with the wavelength of 830 nm. It leads to the reduction of TNF-α concentration in the supernatant and enhancement of cell proliferation. Endothelial cells play an important role in the pathogenesis of diabetes; however, a small number of studies evaluate an impact of LLLT on these cells under conditions of hyperglycemia. Further work on this subject is warranted.


Asunto(s)
Células Endoteliales/efectos de la radiación , Hiperglucemia/radioterapia , Interleucina-6/efectos de la radiación , Terapia por Luz de Baja Intensidad/métodos , Factor de Necrosis Tumoral alfa/efectos de la radiación , Línea Celular , Proliferación Celular/efectos de la radiación , Citocinas/efectos de la radiación , Humanos , Venas Umbilicales
12.
Int J Mol Sci ; 17(1)2015 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-26703588

RESUMEN

Plant phenolic compounds are common dietary antioxidants that possess antioxidant and anti-inflammatory properties. Flaxseed (FS) has been reported to be radioprotective in murine models of oxidative lung damage. Flaxseed's protective properties are attributed to its main biphenolic lignan, secoisolariciresinol diglucoside (SDG). SDG is a free radical scavenger, shown in cell free systems to protect DNA from radiation-induced damage. The objective of this study was to investigate the in vitro radioprotective efficacy of SDG in murine lung cells. Protection against irradiation (IR)-induced DNA double and single strand breaks was assessed by γ-H2AX labeling and alkaline comet assay, respectively. The role of SDG in modulating the levels of cytoprotective enzymes was evaluated by qPCR and confirmed by Western blotting. Additionally, effects of SDG on clonogenic survival of irradiated cells were evaluated. SDG protected cells from IR-induced death and ameliorated DNA damage by reducing mean comet tail length and percentage of γ-H2AX positive cells. Importantly, SDG significantly increased gene and protein levels of antioxidant HO-1, GSTM1 and NQO1. Our results identify the potent radioprotective properties of the synthetic biphenolic SDG, preventing DNA damage and enhancing the antioxidant capacity of normal lung cells; thus, rendering SDG a potential radioprotector against radiation exposure.


Asunto(s)
Antioxidantes/farmacología , Butileno Glicoles/farmacología , Daño del ADN , Células Endoteliales/efectos de los fármacos , Lino/química , Rayos gamma , Glucósidos/farmacología , Pulmón/citología , Extractos Vegetales/farmacología , Animales , Muerte Celular , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/efectos de la radiación , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo
13.
Photochem Photobiol Sci ; 14(5): 972-81, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25793654

RESUMEN

Photodynamic therapy (PDT) has been successfully implemented as a treatment for wet age-related macular degeneration (AMD), but very few photosensitizers have been developed for clinical use. Herein, we describe a novel formulation of liposomal hypocrellin B (LHB) that was prepared by high-pressure homogenization. The encapsulation efficiency and PDT efficacy in vitro of this new preparation were found to remain nearly constant over 1 year. Moreover, LHB is rapidly cleared from the blood, with a half-life of 2.319 ± 0.462 h and a very low serum concentration at 24 h after injection. Testing in a rat model of choroidal neovascularization (CNV) showed that leakage of blood vessels in CNV lesions was significantly reduced when LHB PDT was given at a dose of 1 mg kg(-1) along with yellow laser irradiation; the damage to the collateral retina and the retinal pigment epithelium was minimal. Skin phototoxicity assays showed that only two of the 200 mice given a 4 mg per kg dose of LHB experienced an inflammatory reaction in the auricle irradiated at 24 h after dosing. These data collectively indicate that LHB may be a safe and effective photosensitizer for vascular-targeted PDT of AMD.


Asunto(s)
Perileno/análogos & derivados , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/administración & dosificación , Quinonas/administración & dosificación , Degeneración Macular Húmeda/terapia , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Neovascularización Coroidal , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Oído/patología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Células Endoteliales/efectos de la radiación , Femenino , Liposomas/síntesis química , Pulmón/irrigación sanguínea , Masculino , Ratones , Microvasos/efectos de los fármacos , Microvasos/fisiología , Microvasos/efectos de la radiación , Tamaño de los Órganos , Perileno/administración & dosificación , Perileno/síntesis química , Perileno/farmacocinética , Perileno/toxicidad , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/farmacocinética , Fármacos Fotosensibilizantes/toxicidad , Quinonas/síntesis química , Quinonas/farmacocinética , Quinonas/toxicidad , Ratas , Retina/efectos de los fármacos , Retina/patología , Retina/efectos de la radiación , Piel/efectos de los fármacos , Piel/patología , Piel/efectos de la radiación , Degeneración Macular Húmeda/patología
14.
Lasers Med Sci ; 30(1): 273-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25231826

RESUMEN

Growth factors as vascular endothelial growth factor (VEGF), produced by the endothelial cells, take an essential part in pathological and physiological angiogenesis. The possibility of angiogenesis modulation by application of laser radiation may contribute to the improvement of its use in this process. Thus, the aim of the study was to investigate the influence of low-level laser therapy (LLLT) on the proliferation of endothelial cells, secretion of VEGF-A and presence of soluble VEGF receptors (sVEGFR-1 and sVEGFR-2) in the medium after in vitro culture. Isolated human umbilical vein endothelial cells (HUVECs) were irradiated using a diode laser at a wavelength of 635 nm and power density of 1,875 mW/cm(2). Depending on radiation energy density, the experiment was conducted in four groups: I 0 J/cm(2) (control group), II 2 J/cm(2), III 4 J/cm(2), and IV 8 J/cm(2). The use of laser radiation wavelength of 635 nm, was associated with a statistically significant increase in proliferation of endothelial cells (p = 0.0041). Moreover, at 635-nm wavelength, all doses of radiation significantly reduced the concentration of sVEGFR-1 (p = 0.0197).


Asunto(s)
Células Endoteliales/efectos de la radiación , Terapia por Luz de Baja Intensidad/métodos , Neovascularización Fisiológica , Técnicas de Cultivo de Célula , Proliferación Celular , Endotelio Vascular , Células Endoteliales de la Vena Umbilical Humana , Humanos , Láseres de Semiconductores , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
15.
Free Radic Biol Med ; 74: 200-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24992837

RESUMEN

We previously showed that 5 mM ascorbate radiosensitized early passage radioresistant glioblastoma multiforme (GBM) cells derived from one patient tumor. Here we investigate the sensitivity of a panel of cell lines to 5 mM ascorbate and 6 Gy ionizing radiation, made up of three primary human GBM cells, three GBM cell lines, a human glial cell line, and primary human vascular endothelial cells. The response of different cells lines to ascorbate and/or radiation was determined by measuring viability, colony-forming ability, generation and repair of double-stranded DNA breaks (DSBs), cell cycle progression, antioxidant capacity and generation of reactive oxygen species. Individually, radiation and ascorbate both decreased viability and clonogenicity by inducing DNA damage, but had differential effects on cell cycle progression. Radiation led to G2/M arrest in most cells whereas ascorbate caused accumulation in S phase, which was moderately associated with poor DSB repair. While high dose ascorbate radiosensitized all cell lines in clonogenic assays, the sensitivity to radiation, high dose ascorbate, and combined treatment varied between cell lines. Normal glial cells were similar to GBM cells with respect to free radical scavenging potential and effect of treatment on DNA damage and repair, viability, and clonogenicity. Both GBM cells and normal cells coped equally poorly with oxidative stress caused by radiation and/or high dose ascorbate, dependent primarily on their antioxidant and DSB repair capacity.


Asunto(s)
Ácido Ascórbico/farmacología , Quimioradioterapia/métodos , Células Endoteliales/fisiología , Glioblastoma/terapia , Neuroglía/fisiología , Fármacos Sensibilizantes a Radiaciones/farmacología , Línea Celular Tumoral , Supervivencia Celular , Ensayo de Unidades Formadoras de Colonias , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Células Endoteliales/efectos de los fármacos , Células Endoteliales/efectos de la radiación , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Glioblastoma/patología , Humanos , Neuroglía/efectos de los fármacos , Neuroglía/efectos de la radiación , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Radiación Ionizante , Especies Reactivas de Oxígeno/metabolismo
16.
Int J Nanomedicine ; 9: 3013-26, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25018630

RESUMEN

PURPOSE: The present study examines the use of an external magnetic field in combination with the disruption of tight junctions to enhance the permeability of iron oxide nanoparticles (IONPs) across an in vitro model of the blood-brain barrier (BBB). The feasibility of such an approach, termed magnetic field enhanced convective diffusion (MFECD), along with the effect of IONP surface charge on permeability, was examined. METHODS: The effect of magnetic field on the permeability of positively (aminosilane-coated [AmS]-IONPs) and negatively (N-(trimethoxysilylpropyl)ethylenediaminetriacetate [EDT]-IONPs) charged IONPs was evaluated in confluent monolayers of mouse brain endothelial cells under normal and osmotically disrupted conditions. RESULTS: Neither IONP formulation was permeable across an intact cell monolayer. However, when tight junctions were disrupted using D-mannitol, flux of EDT-IONPs across the bEnd.3 monolayers was 28%, increasing to 44% when a magnetic field was present. In contrast, the permeability of AmS-IONPs after osmotic disruption was less than 5%. The cellular uptake profile of both IONPs was not altered by the presence of mannitol. CONCLUSIONS: MFECD improved the permeability of EDT-IONPs through the paracellular route. The MFECD approach favors negatively charged IONPs that have low affinity for the brain endothelial cells and high colloidal stability. This suggests that MFECD may improve IONP-based drug delivery to the brain.


Asunto(s)
Barrera Hematoencefálica/química , Barrera Hematoencefálica/efectos de la radiación , Electroporación/métodos , Células Endoteliales/química , Células Endoteliales/efectos de la radiación , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/efectos de la radiación , Animales , Línea Celular , Convección , Difusión/efectos de la radiación , Magnetoterapia/métodos , Campos Magnéticos , Ratones
17.
J Nanosci Nanotechnol ; 14(7): 5347-54, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24758030

RESUMEN

Tumor-associated lymphatics and lymphangiogenesis have been shown to play important roles in promoting tumor growth and metastasis. However, the lymphatic system has received much less attention as a target of intervention in cancer treatment compared to the blood vascular system. In this study, we explored the feasibility of photothermal therapy targeting the lymphatic system as a strategy for inhibiting lymphatics-mediated tumor metastasis. Specifically, photothermolysis of lymphatic endothelial cells (LECs) via gold nanoshell-mediated hyperthermia was investigated. Near-Infrared-absorbing Au nanoshells (AuNSs) were synthesized and used as the photothermal coupling agent. After 24-hr incubation, significant amount of the AuNSs were taken up by murine simian virus lymphatic endothelial cells with minimal cytotoxicity. Thermally-induced injury to LECs was found to occur above a threshold temperature of 46 degrees C. Preliminary data also suggested apoptosis as the mechanism of thermally-induced cell death in this temperature range. In a proof-of-concept experiment, AuNS-mediated photothermal heating was found to induce cell death in statistically higher percent of LECs incubated with AuNSs after 15-min laser irradiation compared to the controls. We believed that the findings in this study represent the first step in developing AuNS-mediated photothermal therapy as a potential strategy to disrupt tumor-associated lymphatics.


Asunto(s)
Células Endoteliales/fisiología , Células Endoteliales/efectos de la radiación , Oro/química , Oro/uso terapéutico , Hipertermia Inducida/métodos , Nanopartículas del Metal/química , Nanopartículas del Metal/efectos de la radiación , Fototerapia/métodos , Animales , Apoptosis/fisiología , Apoptosis/efectos de la radiación , Línea Celular , Supervivencia Celular/fisiología , Supervivencia Celular/efectos de la radiación , Células Endoteliales/citología , Luz , Ratones , Dosis de Radiación
18.
Radiat Res ; 181(5): 464-70, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24754562

RESUMEN

Metformin, a biguanide drug used in the treatment of type II diabetes, was evaluated alone and in combination with amifostine, captopril, MESNA or N-acetyl-cysteine (NAC) for its ability to protect when administered 24 h after irradiation. Mouse embryo fibroblasts (MEF), human microvascular endothelial cells (HMEC) and SA-NH mouse sarcoma cells were exposed to 4 Gy in vitro. C3H mice were exposed to 7 Gy and evaluated utilizing an endogenous spleen colony assay system. Amifostine and WR1065, administered 30 min prior to irradiation, were used as positive controls. Treatment of MEF, HMEC and SA-NH cells with metformin elevated survival levels by 1.4-, 1.5- and 1.3-fold compared to 1.9-, 1.8- and 1.6-fold for these same cells treated with WR1065, respectively. Metformin (250 mg/kg) was effective in protecting splenic cells from a 7 Gy dose in vivo (protection factor = 1.8). Amifostine (400 mg/kg), administered 30 min prior to irradiation resulted in a 2.6-fold survival elevation, while metformin administered 24 h after irradiation in combination with NAC (400 mg/kg), MESNA (300 mg/kg) or captopril (200 mg/kg) enhanced survival by 2.6-, 2.8- and 2.4-fold, respectively. Each of these agents has been approved by the FDA for human use and each has a well characterized human safety profile. Metformin alone or in combination with selected sulfhydryl agents possesses radioprotective properties when administered 24 h after radiation exposure comparable to that observed for amifostine administered 30 min prior to irradiation making it a potentially useful agent for radiation countermeasures use.


Asunto(s)
Síndrome de Radiación Aguda/prevención & control , Metformina/uso terapéutico , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/uso terapéutico , Compuestos de Sulfhidrilo/uso terapéutico , Acetilcisteína/administración & dosificación , Acetilcisteína/uso terapéutico , Síndrome de Radiación Aguda/patología , Amifostina/administración & dosificación , Amifostina/farmacología , Amifostina/uso terapéutico , Animales , Captopril/administración & dosificación , Captopril/farmacología , Captopril/uso terapéutico , Línea Celular Transformada , Línea Celular Tumoral , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Células Endoteliales/efectos de los fármacos , Células Endoteliales/efectos de la radiación , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Depuradores de Radicales Libres/administración & dosificación , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/uso terapéutico , Humanos , Mesna/administración & dosificación , Mesna/farmacología , Mesna/uso terapéutico , Metformina/administración & dosificación , Metformina/farmacología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/patología , Protectores contra Radiación/administración & dosificación , Protectores contra Radiación/farmacología , Sarcoma/patología , Compuestos de Sulfhidrilo/administración & dosificación , Compuestos de Sulfhidrilo/farmacología
19.
Mediators Inflamm ; 2012: 498373, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23093820

RESUMEN

Edelweiss (Leontopodium alpinum Cass.) is traditionally employed in folk medicine as an anti-inflammatory remedy. In nature, the plant is sparsely available and protected; therefore production of callus cultures was established. A concentrated ethanolic extract of culture homogenate, with leontopodic acid representing 55 ± 2% of the total phenolic fraction (ECC55), was characterized for anti-inflammatory properties in primary human keratinocytes (PHKs) and endotheliocytes (HUVECs). Inflammatory responses were induced by UVA+UVB, lipopolysaccharide (LPS), oxidized low-density lipoprotein (oxLDL), and a mixture of proinflammatory cytokines. Trichostatin A, a sirtuin inhibitor, was used to induce keratinocyte inflammatory senescence. ECC55 (10-50 µg/mL) protected PHK from solar UV-driven damage, by enhancing early intracellular levels of nitric oxide, although not affecting UV-induced expression of inflammatory genes. Comparison of the dose-dependent inhibition of chemokine (IL-8, IP-10, MCP-1) and growth factor (GM-CSF) release from PHK activated by TNFα + IFNγ showed that leontopodic acid was mainly responsible for the inhibitory effects of ECC55. Sirtuin-inhibited cell cycle, proliferation, and apoptosis markers were restored by ECC55. The extract inhibited LPS-induced IL-6 and VCAM1 genes in HUVEC, as well as oxLDL-induced selective VCAM1 overexpression. Conclusion. Edelweiss cell cultures could be a valuable source of anti-inflammatory substances potentially applicable for chronic inflammatory skin diseases and bacterial and atherogenic inflammation.


Asunto(s)
Antiinflamatorios/farmacología , Asteraceae/química , Células Endoteliales/efectos de los fármacos , Etanol/química , Queratinocitos/efectos de los fármacos , Extractos Vegetales/farmacología , Antiinflamatorios/química , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/efectos de la radiación , Humanos , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Lipoproteínas LDL/farmacología , Extractos Vegetales/química , Rayos Ultravioleta
20.
Photomed Laser Surg ; 30(6): 315-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22506550

RESUMEN

OBJECTIVE: After endothelial cells were ablated by neodymium:yttrium-aluminum-garnet (Nd:YAG) laser irradiation, we investigated the response of pulp cells by examining the expression of transforming growth factor beta-1 (TGF-ß1). BACKGROUND DATA: The reaction of stimulated blood vessels is related to the initiation of dentinogenesis. After artificial injury of endothelial cells, pulp cells migrate to the site of the injured endothelial cells. MATERIALS AND METHODS: Rat aortic endothelial cells were cultured in the lower compartment of the experimental assembly, and a pulsed Nd:YAG laser was used to ablate these cells. Pulp cells were fluorescence labeled and cultured in the upper compartment. After 7-14 days of laser irradiation, total RNA was extracted from the cells in the lower chamber, and RT-PCR was performed to examine the expression of TGF-ß1 and osteocalcin mRNA. TGF-ß1 was also examined with immunohistochemistry. RESULTS: Seven days after laser irradiation, migrating pulp cells that expressed TGF-ß1 were observed in the lower compartment, and the expression of TGF-ß1 mRNA and osteocalcin mRNA was altered. Without laser irradiation, few migrating pulp cells were observed, and the expression of TGF-ß1 mRNA and osteocalcin mRNA was weak. These results suggested that TGF-ß1 mRNA expression is detected earlier in pulp cells rather than in endothelial cells following injury to endothelial cells. CONCLUSIONS: Using the Nd:YAG laser as an ablative stimulant, this study model was useful for investigating pulp-endothelial cell interactions in reparative dentinogenesis.


Asunto(s)
Aluminio/uso terapéutico , Pulpa Dental/efectos de la radiación , Terapia por Luz de Baja Intensidad/métodos , Osteocalcina/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Itrio/uso terapéutico , Animales , Movimiento Celular/fisiología , Movimiento Celular/efectos de la radiación , Células Cultivadas , Pulpa Dental/metabolismo , Dentinogénesis/genética , Dentinogénesis/fisiología , Células Endoteliales/metabolismo , Células Endoteliales/efectos de la radiación , Inmunohistoquímica , Técnicas In Vitro , Masculino , Modelos Animales , Osteocalcina/genética , ARN Mensajero/análisis , Distribución Aleatoria , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA